Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 23(10)2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35628401

RESUMEN

Paired box 9 (PAX9) is a transcription factor of the PAX family functioning as both a transcriptional activator and repressor. Its functional roles in the embryonic development of various tissues and organs have been well studied. However, its roles and molecular mechanisms in cancer development are largely unknown. Here, we review the current understanding of PAX9 expression, upstream regulation of PAX9, and PAX9 downstream events in cancer development. Promoter hypermethylation, promoter SNP, microRNA, and inhibition of upstream pathways (e.g., NOTCH) result in PAX9 silencing or downregulation, whereas gene amplification and an epigenetic axis upregulate PAX9 expression. PAX9 may contribute to carcinogenesis through dysregulation of its transcriptional targets and related molecular pathways. In summary, extensive studies on PAX9 in its cellular and tissue contexts are warranted in various cancers, in particular, HNSCC, ESCC, lung cancer, and cervical SCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Factor de Transcripción PAX9 , Neoplasias de Cabeza y Cuello/genética , Humanos , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Regiones Promotoras Genéticas , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Factores de Transcripción/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1868(9): 166428, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35533906

RESUMEN

Aberrant DNA hypermethylation is associated with oral carcinogenesis. Procaine, a local anesthetic, is a DNA methyltransferase (DNMT) inhibitor that activates anticancer mechanisms. However, its effect on silenced tumor suppressor gene (TSG) activation and its biological role in oral squamous cell carcinoma (OSCC) remain unknown. Here, we report procaine inhibited DNA methylation by suppressing DNMT activity and increased the expression of PAX9, a differentiation gene in OSCC cells. Interestingly, the reactivation of PAX9 by procaine found to inhibit cell growth and trigger apoptosis in OSCC in vitro and in vivo. Likely, the enhanced PAX9 expression after exposure to procaine controls stemness and differentiation through the autophagy-dependent pathway in OSCC cells. PAX9 inhibition abrogated procaine-induced apoptosis, autophagy, and inhibition of stemness. In OSCC cells, procaine improved anticancer drug sensitivity through PAX9, and its deficiency significantly blunted the anticancer drug sensitivity mediated by procaine. Additionally, NRF2 activation by procaine facilitated the antitumor response of PAX9, and pharmacological inhibition of NRF2 by ML385 reduced death and prevented the decrease in the orosphere-forming potential of OSCC cells. Furthermore, procaine promoted antitumor activity in FaDu xenografts in athymic nude mice, and immunohistochemistry data showed that PAX9 expression was significantly enhanced in the procaine group compared to the vehicle control. In conclusion, PAX9 reactivation in response to DNMT inhibition could trigger a potent antitumor mechanism to provide a new therapeutic strategy for OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , ADN , Humanos , Metiltransferasas , Ratones , Ratones Desnudos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Factor 2 Relacionado con NF-E2 , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Procaína/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello
3.
Biochem Biophys Res Commun ; 598: 74-80, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35151207

RESUMEN

The histone methyltransferase SET domain bifurcated 1 (SETDB1) catalyzes the trimethylation of lysine 9 of histone H3, thereby regulating gene expression. In this study, we used conditional knockout mice, where Setdb1 was deleted only in neural crest cells (Setdb1fl/fl,Wnt1-Cre + mice), to clarify the role of SETDB1 in palatal development. Setdb1fl/fl,Wnt1-Cre + mice died shortly after birth due to a cleft palate with full penetration. Reduced palatal mesenchyme proliferation was seen in Setdb1fl/fl,Wnt1-Cre + mice, which might be a possible mechanism of cleft palate development. Quantitative RT-PCR and in situ hybridization showed that expression of the Pax9, Bmp4, Bmpr1a, Wnt5a, and Fgf10 genes, known to be important for palatal development, were markedly decreased in the palatal mesenchyme of Setdb1fl/fl,Wnt1-Cre + mice. Along with these phenomena, SMAD1/5/9 phosphorylation was decreased by the loss of Setdb1. Our results demonstrated that SETDB1 is indispensable for palatal development partially through its proliferative effect. Taken together with previous reports that PAX9 regulates BMP signaling during palatal development which implies that loss of Setdb1 may be involved in the cleft palate development by decreasing SMAD-dependent BMP signaling through Pax9.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/fisiología , Hueso Paladar/embriología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular/genética , Fisura del Paladar/genética , N-Metiltransferasa de Histona-Lisina/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Cresta Neural/fisiopatología , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Hueso Paladar/anomalías , Hueso Paladar/patología , Proteínas Smad/genética , Proteínas Smad/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Kaohsiung J Med Sci ; 38(4): 357-366, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34931758

RESUMEN

To investigate the effect of PAX9 on the progression of cervical cancer (CC). PAX9 expression was quantified in CC tissues and adjacent normal tissues, as well as human CC cell lines and human cervical epithelial cells (HCerEpiC). PAX9-overexpression lentiviral vectors were transfected into CC cell lines, followed by the measurement of proliferation and apoptosis and the quantification of apoptosis-related proteins. In vivo, mice were subcutaneously injected with CaSki cells transfected with PAX9-overexpression lentiviral vectors and control vectors. Then, the volume and weight of tumors were measured followed by hematoxylin and eosin (HE) staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and immunohistochemistry. PAX9 expression in the CC tissues was lower than that in the adjacent normal tissues, which was correlated with the FIGO stage, tumor size, infiltration depth, parametrium invasion, lympho-vascular space invasion tumor-positive lymph nodes, and prognosis. Furthermore, PAX9 in CC cell lines was also lower than in HCerEpiC. PAX9 inhibits the CC cell proliferation and promotes the apoptosis, with the up-regulations of caspase-3, poly(ADP-ribose) polymerase (PARP), and Bax and the down-regulation of Bcl-2. In vivo experiments demonstrated that in the PAX9 group, the tumor weight and volume were lower than those in the vector group accompanying the decreased Ki-67, cleaved-caspase-3, and Bax expressions and the increased TUNEL and Bcl-2 expression. PAX9 was lowly expressed in the CC tissues and associated with the clinicopathological characteristics and prognosis. PAX9 could inhibit proliferation of CC cell lines and promote the apoptosis, thus suppressing the tumor growth in vivo, indicating its potential therapeutic role for CC treatment.


Asunto(s)
Genes Supresores de Tumor , Factor de Transcripción PAX9 , Neoplasias del Cuello Uterino , Animales , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Neoplasias del Cuello Uterino/patología
5.
Cancer Res ; 81(18): 4696-4708, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34341073

RESUMEN

Abnormalities in genetic and epigenetic modifications can lead to drastic changes in gene expression profiles that are associated with various cancer types. Small cell lung cancer (SCLC) is an aggressive and deadly form of lung cancer with limited effective therapies currently available. By utilizing a genome-wide CRISPR-Cas9 dropout screen in SCLC cells, we identified paired box protein 9 (PAX9) as an essential factor that is overexpressed in human malignant SCLC tumor samples and is transcriptionally driven by the BAP1/ASXL3/BRD4 epigenetic axis. Genome-wide studies revealed that PAX9 occupies distal enhancer elements and represses gene expression by restricting enhancer activity. In multiple SCLC cell lines, genetic depletion of PAX9 led to significant induction of a primed-active enhancer transition, resulting in increased expression of a large number of neural differentiation and tumor-suppressive genes. Mechanistically, PAX9 interacted and cofunctioned with the nucleosome remodeling and deacetylase (NuRD) complex at enhancers to repress nearby gene expression, which was reversed by pharmacologic HDAC inhibition. Overall, this study provides mechanistic insight into the oncogenic function of the PAX9/NuRD complex epigenetic axis in human SCLC and suggests that reactivation of primed enhancers may have potential therapeutic efficacy in treating SCLC expressing high levels of PAX9. SIGNIFICANCE: A genome-wide screen in small cell lung cancer reveals PAX9/NuRD-mediated epigenetic enhancer silencing and tumor progression, supporting the development of novel personalized therapeutic approaches targeting the PAX9-regulated network.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/metabolismo , Factor de Transcripción PAX9/metabolismo , Animales , Sistemas CRISPR-Cas , Diferenciación Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Modelos Biológicos , Neoplasias/patología , Factor de Transcripción PAX9/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Factores de Transcripción/genética
6.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188561, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33965511

RESUMEN

Paired box 9 (PAX9) gene belongs to the PAX family, which encodes a family of metazoan transcription factors documented by a conserved DNA binding paired domain 128-amino-acids, critically essential for physiology and development. It is primarily expressed in embryonic tissues, such as the pharyngeal pouch endoderm, somites, neural crest-derived mesenchyme, and distal limb buds. PAX9 plays a vital role in craniofacial development by maintaining the odontogenic potential, mutations, and polymorphisms associated with the risk of tooth agenesis, hypodontia, and crown size in dentition. The loss-of-function of PAX9 in the murine model resulted in a short life span due to the arrest of cleft palate formation and skeletal abnormalities. According to recent studies, the PAX9 gene has a significant role in maintaining squamous cell differentiation, odontoblast differentiation of pluripotent stem cells, deregulation of which is associated with tumor initiation, and malignant transformation. Moreover, PAX9 contributes to promoter hypermethylation and alcohol- induced oro-esophageal squamous cell carcinoma mediated by downregulation of differentiation and apoptosis. Likewise, PAX9 activation is also reported to be associated with drug sensitivity. In summary, this current review aims to understand PAX9 function in the regulation of development, differentiation, and carcinogenesis, along with the underlying signaling pathways for possible cancer therapeutics.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción PAX9/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Mutación , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Células Madre Neoplásicas/patología , Organogénesis , Factor de Transcripción PAX9/genética , Polimorfismo de Nucleótido Simple , Transducción de Señal
7.
J Pathol ; 253(4): 384-395, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33314197

RESUMEN

Alcohol drinking has been established as a major risk factor for esophageal diseases. Our previous study showed that ethanol exposure inhibited PAX9 expression in human esophageal squamous epithelial cells in vitro and in vivo. In this study, we aimed to investigate the molecular pathways through which alcohol drinking suppresses PAX9 in esophageal squamous epithelial cells. We first demonstrated the inhibition of NOTCH by ethanol exposure in vitro. NOTCH regulated PAX9 expression in KYSE510 and KYSE410 cells in vitro and in vivo. RBPJ and NOTCH intracellular domain (NIC) D1 ChIP-PCR confirmed Pax9 as a direct downstream target of NOTCH signaling in mouse esophagus. NOTCH inhibition by alcohol drinking was further validated in mouse esophagus and human tissue samples. In conclusion, ethanol exposure inhibited NOTCH signaling and thus suppressed PAX9 expression in esophageal squamous epithelial cells in vitro and in vivo. Our data support a novel mechanism of alcohol-induced esophageal injury through the inhibition of NOTCH-PAX9 signaling. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Carcinoma de Células Escamosas de Esófago/patología , Factor de Transcripción PAX9/efectos de los fármacos , Receptores Notch/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Etanol/toxicidad , Humanos , Ratones , Factor de Transcripción PAX9/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo
8.
Mol Biol Evol ; 38(5): 1860-1873, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33355664

RESUMEN

Eutherian dentition has been the focus of a great deal of studies in the areas of evolution, development, and genomics. The development of molar teeth is regulated by an antero-to-posterior cascade mechanism of activators and inhibitors molecules, where the relative sizes of the second (M2) and third (M3) molars are dependent of the inhibitory influence of the first molar (M1). Higher activator/inhibitor ratios will result in higher M2/M1 or M3/M1. Pax9 has been shown to play a key role in tooth development. We have previously shown that a G-quadruplex in the first intron of Pax9 can modulate the splicing efficiency. Using a sliding window approach with we analyzed the association of the folding energy (Mfe) of the Pax9 first intron with the relative molar sizes in 42 mammalian species, representing 9 orders. The Mfe of two regions located in the first intron of Pax9 were shown to be significantly associated with the M2/M1 and M3/M1 areas and mesiodistal lengths. The first region is located at the intron beginning and can fold into a stable G4 structure, whereas the second is downstream the G4 and 265 bp from intron start. Across species, the first intron of Pax9 varied in G-quadruplex structural stability. The correlations were further increased when the Mfe of the two sequences were added. Our results indicate that this region has a role in the evolution of the mammalian dental pattern by influencing the relative size of the molars.


Asunto(s)
Evolución Biológica , Euterios/anatomía & histología , Diente Molar/anatomía & histología , Factor de Transcripción PAX9/metabolismo , Animales , Euterios/metabolismo , G-Cuádruplex , Intrones
10.
Development ; 147(21)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32467233

RESUMEN

Nonsyndromic clefts of the lip and palate are common birth defects resulting from gene-gene and gene-environment interactions. Mutations in human MSX1 have been linked to orofacial clefting and we show here that Msx1 deficiency causes a growth defect of the medial nasal process (Mnp) in mouse embryos. Although this defect alone does not disrupt lip formation, Msx1-deficient embryos develop a cleft lip when the mother is transiently exposed to reduced oxygen levels or to phenytoin, a drug known to cause embryonic hypoxia. In the absence of interacting environmental factors, the Mnp growth defect caused by Msx1 deficiency is modified by a Pax9-dependent 'morphogenetic regulation', which modulates Mnp shape, rescues lip formation and involves a localized abrogation of Bmp4-mediated repression of Pax9 Analyses of GWAS data revealed a genome-wide significant association of a Gene Ontology morphogenesis term (including assigned roles for MSX1, MSX2, PAX9, BMP4 and GREM1) specifically for nonsyndromic cleft lip with cleft palate. Our data indicate that MSX1 mutations could increase the risk for cleft lip formation by interacting with an impaired morphogenetic regulation that adjusts Mnp shape, or through interactions that inhibit Mnp growth.


Asunto(s)
Hipoxia/embriología , Hipoxia/metabolismo , Labio/embriología , Factor de Transcripción MSX1/deficiencia , Morfogénesis , Animales , Proteína Morfogenética Ósea 4/metabolismo , Labio Leporino/embriología , Labio Leporino/genética , Labio Leporino/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genoma , Proteínas de Homeodominio/metabolismo , Humanos , Hipoxia/genética , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Morfogénesis/genética , Mutación/genética , Nariz/embriología , Oxígeno/metabolismo , Factor de Transcripción PAX9/metabolismo , Fenitoína , Respiración , Regulación hacia Arriba/genética
11.
Dev Biol ; 458(2): 246-256, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31765609

RESUMEN

In this study, we investigated the role of the transcription factor Six2 in palate development. Six2 was selected using the SysFACE tool to predict genes from the 2p21 locus, a region associated with clefting in humans by GWAS, that are likely to be involved in palatogenesis. We functionally validated the predicted role of Six2 in palatogenesis by showing that 22% of Six2 null embryos develop cleft palate. Six2 contributes to palatogenesis by promoting mesenchymal cell proliferation and regulating bone formation. The clefting phenotype in Six2-/- embryos is similar to Pax9 null embryos, so we examined the functional relationship of these two genes. Mechanistically, SIX2 binds to a PAX9 5' upstream regulatory element and activates PAX9 expression. In addition, we identified a human SIX2 coding variant (p.Gly264Glu) in a proband with cleft palate. We show this missense mutation affects the stability of the SIX2 protein and leads to decreased PAX9 expression. The low penetrance of clefting in the Six2 null mouse combined with the mutation in one patient with cleft palate underscores the potential combinatorial interactions of other genes in clefting. Our study demonstrates that Six2 interacts with the developmental gene regulatory network in the developing palate.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Transcripción PAX9/genética , Factores de Transcripción/metabolismo , Animales , Fisura del Paladar/embriología , Fisura del Paladar/genética , Anomalías Craneofaciales/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Morfogénesis , Proteínas del Tejido Nervioso/metabolismo , Osteogénesis , Factor de Transcripción PAX9/metabolismo , Factores de Transcripción Paired Box , Hueso Paladar/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética
12.
Development ; 146(18)2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444215

RESUMEN

Developmental defects affecting the heart and aortic arch arteries are a significant phenotype observed in individuals with 22q11 deletion syndrome and are caused by a microdeletion on chromosome 22q11. TBX1, one of the deleted genes, is expressed throughout the pharyngeal arches and is considered a key gene, when mutated, for the arch artery defects. Pax9 is expressed in the pharyngeal endoderm and is downregulated in Tbx1 mutant mice. We show here that Pax9-deficient mice are born with complex cardiovascular malformations that affect the outflow tract and aortic arch arteries with failure of the 3rd and 4th pharyngeal arch arteries to form correctly. Transcriptome analysis indicated that Pax9 and Tbx1 may function together, and mice double heterozygous for Tbx1/Pax9 presented with a significantly increased incidence of interrupted aortic arch when compared with Tbx1 heterozygous mice. Using a novel Pax9Cre allele, we demonstrated that the site of this Tbx1-Pax9 genetic interaction is the pharyngeal endoderm, therefore revealing that a Tbx1-Pax9-controlled signalling mechanism emanating from the pharyngeal endoderm is required for crucial tissue interactions during normal morphogenesis of the pharyngeal arch artery system.


Asunto(s)
Arterias/embriología , Región Branquial/irrigación sanguínea , Sistema Cardiovascular/embriología , Endodermo/embriología , Morfogénesis , Factor de Transcripción PAX9/metabolismo , Faringe/embriología , Proteínas de Dominio T Box/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Diferenciación Celular/genética , Embrión de Mamíferos/anomalías , Eliminación de Gen , Redes Reguladoras de Genes , Heterocigoto , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Cresta Neural/patología , Factor de Transcripción PAX9/deficiencia , Unión Proteica , Transducción de Señal
13.
PLoS One ; 13(9): e0202747, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30208064

RESUMEN

Snail2 is a zinc-finger transcription factor best known to repress expression of genes encoding cell adherence proteins to facilitate induction of the epithelial-to-mesenchymal transition. While this role has been best documented in the developmental migration of the neural crest and mesoderm, here we expand on previously reported preliminary findings that morpholino knock-down of snai2 impairs the generation of hematopoietic stem cells (HSCs) during zebrafish development. We demonstrate that snai2 morphants fail to initiate HSC specification and show defects in the somitic niche of migrating HSC precursors. These defects include a reduction in sclerotome markers as well as in the Notch ligands dlc and dld, which are known to be essential components of HSC specification. Accordingly, enforced expression of the Notch1-intracellular domain was capable of rescuing HSC specification in snai2 morphants. To parallel our approach, we obtained two mutant alleles of snai2. In contrast to the morphants, homozygous mutant embryos displayed no defects in HSC specification or in sclerotome development, and mutant fish survive into adulthood. However, when these homozygous mutants were injected with snai2 morpholino, HSCs were improperly specified. In summary, our morpholino data support a role for Snai2 in HSC development, whereas our mutant data suggest that Snai2 is dispensable for this process. Together, these findings further support the need for careful consideration of both morpholino and mutant phenotypes in studies of gene function.


Asunto(s)
Factores de Transcripción de la Familia Snail/genética , Proteínas de Pez Cebra/genética , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Embrión no Mamífero/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Morfolinos/metabolismo , Mutagénesis Sitio-Dirigida , Factor de Transcripción PAX9/metabolismo , Fenotipo , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Factores de Transcripción de la Familia Snail/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo
14.
J Pathol ; 244(2): 164-175, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29055049

RESUMEN

PAX9 is a transcription factor of the PAX family characterized by a DNA-binding paired domain. Previous studies have suggested a potential role of PAX9 in squamous cell differentiation and carcinogenesis of the oro-oesophageal epithelium. However, its functional roles in differentiation and carcinogenesis remain unclear. In this study, Pax9 deficiency in mouse oesophagus promoted cell proliferation, delayed cell differentiation, and altered the global gene expression profile. Ethanol exposure downregulated PAX9 expression in human oesophageal epithelial cells in vitro and mouse forestomach and tongue in vivo. We further showed that PAX9 was downregulated in human oro-oesophageal squamous cell carcinoma (OESCC), and its downregulation was associated with alcohol drinking and promoter hypermethylation. Moreover, ad libitum feeding with a liquid diet containing ethanol for 40 weeks or Pax9 deficiency promoted N-nitrosomethylbenzylamine-induced squamous cell carcinogenesis in mouse tongue, oesophagus, and forestomach. In conclusion, PAX9 regulates squamous cell differentiation in the oro-oesophageal epithelium. Alcohol drinking and promoter hypermethylation are associated with PAX9 silencing in human OESCC. PAX9 downregulation may contribute to alcohol-associated oro-oesophageal squamous cell carcinogenesis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Factor de Transcripción PAX9/metabolismo , Factores de Transcripción Paired Box/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Neoplasias de la Lengua/metabolismo , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Metilación de ADN , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción PAX9/genética , Factores de Transcripción Paired Box/deficiencia , Factores de Transcripción Paired Box/genética , Regiones Promotoras Genéticas , Factores de Riesgo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/patología , Transcriptoma
15.
J Dent Res ; 96(11): 1282-1289, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28813171

RESUMEN

To date, surgical interventions are the only means by which craniofacial anomalies can be corrected so that function, esthetics, and the sense of well-being are restored in affected individuals. Unfortunately, for patients with cleft palate-one of the most common of congenital birth defects-treatment following surgery is prolonged over a lifetime and often involves multidisciplinary regimens. Hence, there is a need to understand the molecular pathways that control palatogenesis and to translate such information for the development of noninvasive therapies that can either prevent or correct cleft palates in humans. Here, we use the well-characterized model of the Pax9-/- mouse, which displays a consistent phenotype of a secondary cleft palate, to test a novel therapeutic. Specifically, we demonstrate that the controlled intravenous delivery of a novel mouse monoclonal antibody replacement therapy, which acts as an agonist for the ectodysplasin (Eda) pathway, can resolve cleft palate defects in Pax9-/- embryos in utero. Such pharmacological interventions did not reverse the arrest in tooth, thymus, and parathyroid gland development, suggesting that the relationship of Pax9 to the Eda/Edar pathway is both unique and essential for palatogenesis. Expression analyses and unbiased gene expression profiling studies offer a molecular explanation for the resolution of palatal defects, showing that Eda and Edar-related genes are expressed in normal palatal tissues and that the Eda/Edar signaling pathway is downstream of Pax9 in palatogenesis. Taken together, our data uncover a unique relationship between Pax9 and the Eda/Edar signaling pathway that can be further exploited for the development of noninvasive, safe, and effective therapies for the treatment of cleft palate conditions and other single-gene disorders affecting the craniofacial complex.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Fisura del Paladar/tratamiento farmacológico , Fisura del Paladar/embriología , Receptor Edar/agonistas , Factor de Transcripción PAX9/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Endogámicos , Morfogénesis , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
16.
Mol Med Rep ; 16(1): 806-816, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28560390

RESUMEN

Abnormal paired box 9 (PAX9) expression is associated with tumorigenesis, cancer development, invasion and metastasis. The present study investigated the prognostic significance of PAX9 in esophageal squamous cell carcinoma (ESCC) and its role in predicting radiation sensitivity. A total of 52.8% (121/229) ESCC tissues were positive for PAX9. The 1­, 3­ and 5­year disease­free survival (DFS) rates were 72.2, 35.2 and 5.6%, respectively, and the overall survival (OS) rates were and 86.1, 44.4, and 23.1%, respectively, in PAX9­positive tumors. In PAX9­negative tumors, the one­, three­ and five­year DFS rates were 76.9, 47.9 and 24.0%, and the OS rates were 90.9, 57.9 and 38.8%, respectively. Univariate analysis revealed that PAX9, differentiation, T stage, lymph node metastasis, and tumor­node­metastasis stage were associated with OS. Multivariate analysis of DFS and OS revealed that the hazard ratios for PAX9 were 0.624 (95% CI: 0.472­0.869, P=0.004) and 0.673 (95% CI: 0.491­0.922, P=0.014), respectively. Patients that received adjuvant therapy exhibited significant differences in the 5­year DFS (P<0.001) and OS (P<0.001). PAX9­positive ESCC patients who received post­surgery radiotherapy had a significantly greater 5­year DFS (P=0.011) and OS (P=0.009) than patients who received surgery only. Thus, PAX9 may be an independent prognostic factor for the surgical treatment of ESCC and a possible predictor of radiation sensitivity.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Factor de Transcripción PAX9/genética , Tolerancia a Radiación/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Quimioradioterapia , Terapia Combinada , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/radioterapia , Carcinoma de Células Escamosas de Esófago , Femenino , Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Factor de Transcripción PAX9/metabolismo , Pronóstico
17.
J Endocrinol ; 231(3): 181-195, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27756766

RESUMEN

PAX2, a member of paired box family, is an essential transcription factor for the organ development in vertebrates including teleosts, yet no evidence has been shown for its involvement in reproduction. To study this, partial- and/or full-length cDNA of pax2 was isolated from the ovary of catfish, Clarias batrachus, along with its other Pax family members, pax1 and pax9 Tissue distribution and ontogeny expression analysis indicated the prevalence of pax2 but not pax1 and pax9 in ovary. Varied phase-wise expression during ovarian cycle and elevation of pax2 after human chorionic gonadotropin induction showed probable regulation by gonadotropins. Pax2 could be localized in various stages of oocytes and in follicular layer of vitellogenic and post-vitellogenic oocytes. To assess the functional significance of pax2, transient RNA silencing was performed using primary catfish ovarian follicle culture, in vitro, and in catfish, in vivo, through ovary-targeted injection of PEI-esiRNA. Pax2 siRNA treatment reduced the expression of various transcripts related to ovarian development like signaling molecules such as wnt4 and wnt5, estrogen receptors, several steroidogenic enzymes and transcription factors. These transitions in transcript levels might have been mediated by Pax2 acting upstream of wnt4/5 that may play a role in steroidogenesis and/or ovarian development along with ad4bp/sf-1 or by direct or indirect interaction with steroidogenic enzyme genes, which is evident from the change in the levels of serum estradiol-17ß but not 17α,20ß-dihydroxy-4-pregnen-3-one. Taken together, it seems that pax2 has a plausible role during ovarian development and/or recrudescence of catfish either directly or indirectly through Wnt signaling pathway.


Asunto(s)
Bagres/crecimiento & desarrollo , Bagres/metabolismo , Proteínas de Peces/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Factor de Transcripción PAX2/metabolismo , Esteroides/biosíntesis , Animales , Bagres/genética , Gonadotropina Coriónica/administración & dosificación , Clonación Molecular , ADN Complementario/genética , Estradiol/metabolismo , Femenino , Proteínas de Peces/antagonistas & inhibidores , Proteínas de Peces/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Hidroxiprogesteronas/metabolismo , Ovario/efectos de los fármacos , Factor de Transcripción PAX2/antagonistas & inhibidores , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Técnicas de Cultivo de Tejidos , Vía de Señalización Wnt
18.
Int. j. morphol ; 33(2): 647-652, jun. 2015. ilus
Artículo en Inglés | LILACS | ID: lil-755523

RESUMEN

Cleft lip and palate (CLP) is a congenital anomaly characterized by the inappropriate fusion of the upper lip, alveolus, and secondary palate. This study investigated whether expression of interferon regulatory fac tor 6 (IRF6), receptor-like tyrosine kinase (RYK), and paired-box 9 (PAX9), which are essential for the normal development and morphogenesis of craniofacial structures, is dysregulated in children with CLP. Oral mucosa tissue samples were obtained from patients with complete bilateral (CB) CLP (n= 19) during corrective plastic surgery and unaffected control subjects (n= 7). IRF6, RYK, and PAX9 expression was assessed by immunohistochemistry, and data were analyzed with the Mann-Whitney test. In patients, IRF6 immunoreactivity in the connective tissue was moderate to high, but the overall number of IRF6-positive oral epithelial cells was lower than that in controls (z= -3.41; P= 0.01). RYK expression was observed only sporadically in the oral epithelium of 4 patients, in contrast to the control group (z= -3.75; P< 0.001). PAX9-positive epithelial cells were present in low to moderate numbers in patients with CBCLP, while an abundance of these cells was observed in the basal layer of the oral epithelium in controls (z= -3.60; P<0.001). IRF6 is the main connective tissue regulatory factor in CBCLP, and its low level of expression in the oral epithelium suggests a reduced potential for epitheliocyte differentiation, while low PAX9 and RYK expression may explain the decreased cell migration and cleft remodeling in CBCLP.


La fisura labial y palatina (FLP) son anomalías congénitas caracterizadas por la fusión inadecuada del labio superior, alvéolo y paladar secundario. En este estudio se investigó si en niños con FLP hay una desregulación de la expresión del factor regulador de interferón 6 (IRF6), del receptor de la tirosina quinasa (RYK), y del factor de transcripción PAX9, que son esenciales para el desarrollo normal y la morfogénesis de las estructuras craneofaciales. Se obtuvieron muestras de la mucosa oral de pacientes con FLP completa bilateral (CB) (n= 19), tomadas durante la realización de cirugía plástica correctiva, y de sujetos de control no afectados (n= 7). Se evaluó la expresión de IRF6, RYK y PAX9 por inmunohistoquímica, y los datos se analizaron con la prueba de Mann-Whitney. En los pacientes, la inmunoreactividad de IRF6 en el tejido conectivo fue de moderada a alta, pero el número total de células epiteliales orales positivas para IRF6 fue menor que en los controles (z= -3,41; P= 0,01). La expresión de RYK se observó sólo esporádicamente en el epitelio oral de 4 pacientes, en contraste con el grupo control (z= -3,75; P<0.001). Células epiteliales positivas para PAX9 estaban presentes en números bajos a moderados en pacientes con FLP completa bilateral, mientras que se observó una abundante cantidad de estas células en la capa basal del epitelio oral en los controles (z= -3,60; P<0,001). IRF6 es el principal factor regulador del tejido conectivo con FLP completa bilateral, y su bajo nivel de expresión en el epitelio oral sugiere un potencial reducido para la diferenciación del epitelio, mientras que la expresión baja de PAX9 y RYK pueden explicar la disminución de la migración celular y la remodelación de la fisura con FLP completa bilateral.


Asunto(s)
Humanos , Masculino , Femenino , Niño , Fisura del Paladar/metabolismo , Fisura del Paladar/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factores Reguladores del Interferón/metabolismo , Factor de Transcripción PAX9/metabolismo , Inmunohistoquímica
19.
J Appl Toxicol ; 35(11): 1390-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25619733

RESUMEN

High levels of homocysteine (Hcy) are related to an increased risk of the occurrence of congenital anomalies, including limb defects. However, few evaluations about how toxic levels of Hcy affect limb development have been reported. We investigated whether Hcy can affect the cell cycle proteins and proteins involved in mesenchymal cell differentiation during limb development, in a chicken embryo model. Embryos were treated with 20 µmol d-l Hcy/50 µl saline at embryonic day 2 and analyzed at embryonic day 6. Untreated control embryos received exclusively 50 µl saline solution. To identify cells in proliferation and cell cycle proteins, as well as Pax1/9 and Sox9 proteins, we performed immunolocalization and flow cytometry analyses using the antibodies anti-phosphohistone H3, anti-p53, anti-p21, anti-proliferating cell nuclear antigen, anti-Pax1, anti-Pax9 and anti-Sox9. No significant differences in cell proliferation were observed between Hcy-treated and untreated embryos. We observed a decrease of the proliferating cell nuclear antigen and p21 proteins, both involved in the G1 phase of cell cycle progression. On the other hand, in mesenchymal cells of the limbs, Hcy induces an increase of p53 protein, which can be activated by DNA damage. In cell differentiation, Hcy induced an increase mainly of Pax9 and Sox9 proteins. Our data indicate that the treatment with Hcy changes the mesenchymal cell dynamics during limb development, but does not change the morphology of the cartilage molds. These findings provide information to understand better the cellular basis of the toxicity of Hcy on chondrogenesis during limb development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Homocisteína/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Organogénesis/efectos de los fármacos , Animales , Embrión de Pollo , Daño del ADN , Extremidades/embriología , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción PAX9/genética , Factor de Transcripción PAX9/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Biomed Res Int ; 2014: 309385, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25197636

RESUMEN

Both Pax1 and Pax9 belong to the important paired box gene family (PAX), which mainly participates in animal development and sclerotome differentiation. To date, the precise molecular mechanism and related signaling pathway of Pax1 remain unclear. In our study, microinjection of morpholino- (MO-) modified antisense oligonucleotides against pax1b induced pectoral fin bud defects. Furthermore, we demonstrate that the phenotypes caused by the knockdown of Pax1b in zebrafish could not be phenocopied by pax9 MO and could not be rescued by either Pax1a or Pax9 overexpression. We further find that Pax1b affects the expression of col2a1, Uncx4.1, Noggin3, and aggrecan, confirming the role of Pax1b in chondrocyte differentiation and bone maturation. Moreover, we identify an interaction between PAX1 and FOXO1 and find that the interaction was enhanced under hypoxia stress. Together, this evidence for cell death caused by pax1b knockdown provides new insight into the role of the Pax protein family in cell fate determination and tissue specification.


Asunto(s)
Aletas de Animales/embriología , Aletas de Animales/metabolismo , Desarrollo Embrionario , Factor de Transcripción PAX9/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Aletas de Animales/anomalías , Animales , Desarrollo Óseo , Muerte Celular , Desarrollo Embrionario/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Morfogénesis , Factor de Transcripción PAX9/antagonistas & inhibidores , Factor de Transcripción PAX9/genética , Factores de Transcripción Paired Box/genética , Fenotipo , Unión Proteica , Estrés Fisiológico , Cola (estructura animal)/anomalías , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...